• Keine Ergebnisse gefunden

Low Ethanol Concentrations Promote Endothelial Progenitor Cell Capacity and Reparative Function

N/A
N/A
Protected

Academic year: 2022

Aktie "Low Ethanol Concentrations Promote Endothelial Progenitor Cell Capacity and Reparative Function"

Copied!
10
0
0

Wird geladen.... (Jetzt Volltext ansehen)

Volltext

(1)

Research Article

Low Ethanol Concentrations Promote Endothelial Progenitor Cell Capacity and Reparative Function

Lars Brodowski,1,2Bianca Schröder-Heurich,1Berina Kipke,1,2Cara Schmidt,1 Constantin S. von Kaisenberg,2and Frauke von Versen-Höynck 1,2

1Gynecology Research Unit, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany

2Department of Obstetrics and Gynecology, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany

Correspondence should be addressed to Frauke von Versen-Höynck; vonversen-hoeynck.frauke@mh-hannover.de Received 24 June 2020; Revised 30 July 2020; Accepted 10 September 2020; Published 22 September 2020

Academic Editor: Victor Garcia

Copyright © 2020 Lars Brodowski et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Background. Endothelial progenitor cells (EPCs) are recruited to injured endothelium and contribute to its regeneration. There is evidence that moderate ethanol consumption prevents the development and progression of atherosclerosis in a variety ofin vitro andin vivomodels and increases the mobilization of progenitor cells. Furthermore, there are studies that identied ethanol at low concentration as a therapeutic tool to mobilize progenitor cells in peripheral blood. At the same time, the cell number of EPCs represents a close link to cardiovascular system constitution and function and contributes to cardiovascular risk. The aim of this study was to evaluate the eect of low dose ethanol on typical features of endothelial colony-forming cells (ECFCs), a proliferative subtype of EPCs.Methods and Results. We tested whether ethanol impacts the functional abilities of ECFC (e.g., migration, tube formation, and proliferation) usingin vitroassays, the intercommunication of ECFC by exploring cell surface molecules by ow cytometry, and the expression of (anti-)angiogenic molecules by ELISA. Low concentrations of ethanol concentration promoted migration, proliferation, and tubule formation of ECFC. The expression of the cell surface marker VE- cadherin, a protein which plays an important role in cell-cell interaction, was enhanced by ethanol, while (anti-)angiogenic molecule expression was not impacted. Conclusion. Ethanol at moderate concentrations increases the angiogenic abilities of endothelial progenitor cells thus possibly contributing to vasoprotection.

1. Introduction

Cardiovascular disease (CVD) is a major cause of death worldwide. Most CVDs are based on atherosclerosis, a degenerative process of the arterial vascular endothelium induced by oxidative stress and chronic inflammatory status.

The classic risk factors include smoking, diabetes mellitus, arterial hypertension, changes in total cholesterol, obesity, and excessive consumption of ethanol. Several studies have shown that strong ethanol consumption in adults correlates with the occurrence of CVD [1–4]. On the contrary, moder- ate ethanol consumption is associated with decreased mor- bidity and mortality from ischemic heart disease [5–7].

The protective effect of ethanol on the cardiovascular system has been attributed to the modulation of blood lipoproteins and to reduced platelet activation and thus diminished formation of thrombi. Furthermore, other

studies suggest that ethanol has a direct protective effect on the myocardium [5, 8, 9].

Tissue regeneration is the focus of therapy in the treat- ment of CVDs. Proper vascular formation is essential in this context. Endothelial progenitor cells (EPCs) and especially their proliferative subtype endothelial colony-forming cells (ECFCs) are of pivotal importance for endothelial homeosta- sis and vascular remodeling. In addition, they represent a promising cell source for the revascularization of damaged tissue. The concept that alterations in EPC biology impact endothelial cell function is supported by recent studies show- ing that decreased numbers of circulating EPCs correlate with impaired endothelial function [10, 11]. Both a reduced number and impaired function of circulating EPCs have been observed in patients with CVD [12]. The number of circulat- ing EPCs predicts the occurrence of cardiovascular events and can help identify patients at increased risk [13]. ECFC

Volume 2020, Article ID 4018478, 10 pages https://doi.org/10.1155/2020/4018478

(2)

can be isolated from cord blood or peripheral adult blood and migrate to sites of vessel formation, possessing the ability to differentiate into mature endothelial cells, to participate in vessel repair, and to form de novo endothelium [14].

Angiogenic proteins and vascular cell adhesion proteins are important determinants of vascular health. Vascular endothelial growth factor (VEGF) has been demonstrated to promote atherosclerotic plaque progression [15]. More- over, VEGF and placental growth factor (PIGF) stimulate endothelial cell proliferation and migration and mediate vascular growth and angiogenesis. VEGF and its soluble receptor soluble fms-like tyrosine kinase-1 (sFlt-1) are impli- cated in vascular damage and repair in CVD [16]. Vascular cell adhesion protein 1 (VCAM), platelet and endothelial cell adhesion molecule 1 (PECAM1), and vascular endothelial cadherin (VE-cadherin) play an important role in leukocyte extravasation, inflammatory processes, and vascular perme- ability and are mainly involved in the homing of cells to sites of endothelial repair and angiogenesis [17].

While in cohort studies moderate ethanol exposure is associated with reduced cardiovascular morbidity [18], little is known about the underlying pathomechanisms, specifi- cally with regard to EPC biology. Therefore, in in vitro models, we investigated whether ethanol contributes to a poor EPC response or rather has a protective cardiovascular effect.

2. Materials and Methods

2.1. ECFC Isolation and Culture. ECFCs from cord blood were isolated as previously described [19]. Briefly, umbilical cord venous blood was collected immediately after delivery into sterile EDTA-coated tubes. Blood samples were centri- fuged within 3 h of collection at 2,000 g for 5 min. Mononu- clear cells (MCs) were isolated by density gradient centrifugation. The plasma was removed for collection and replaced with the same volume of plasma replacement buffer consisting of phosphate-buffered saline solution (PBS) sup- plemented with 0.025 M EDTA (Sigma-Aldrich, Steinheim, Germany or St. Louis, MO) and 1% (v/v) penicillin/strepto- mycin (Sigma-Aldrich). The sample volume was doubled by adding isolation buffer (PBS, 2% (v/v), fetal bovine serum (FBS, Biochrom KG, Berlin, Germany or Life Technologies, Carlsbad, CA), and 1% penicillin/streptomycin), and the sample was gently mixed. Samples were layered on Ficoll Plus (GE Healthcare, Buckinghamshire, England or Piscataway, NJ) and spun at 400 g for 40 min in a swinging bucket centri- fuge with a brake in the off position. The MC fraction was collected and washed two times with an isolation buffer.

Mononuclear cells were cultured in endothelial growth medium 2 (EGM-2, Lonza, Basel, Switzerland or Walkers- ville, MD), supplemented with supplier-recommended con- centrations of human recombinant epidermal growth factor, VEGF, ascorbic acid, hydrocortisone, heparin, and recombinant insulin-like growth factor, 10% FBS and 1%

penicillin/streptomycin. The MCs were plated at a density of 5 × 107 cells/well on collagen-coated 6-well plates (BD Biosciences, Heidelberg, Germany, or Billerica, MA) and incubated at 37°C, 5% CO2. The medium was changed daily

for 10 days and then every second day. Thefirst appearance of ECFC colonies was noted as well-circumscribed mono- layers of >50 rapidly proliferating, cobblestone-appearing cells. Colonies were identified by visual inspection using an inverted microscope (Olympus, Tokyo, Japan; Zeiss, Thornwood, NY). Well-defined colonies were released from the plates using cloning rings and trypsin-EDTA and col- lected. The cells from each separate colony were placed into a well of a collagen-coated 6-well plate and after becoming 80–90% confluent, subsequently passaged into collagen- coated T25 cultureflasks. After reaching 80–90% confluence, the cells in the T25flasks were passaged into gelatin-coated T75flasks. At 80–90% confluence, these cells were harvested, phenotyped, and frozen in a freezing medium containing 92% FCS and 8% DMSO (Sigma-Aldrich, Steinheim, Germany).

Flow cytometric analyses to confirm the ECFC pheno- type were performed using surface markers CD31, CD34, CD133, VEGFR-2, and CD45 as well as appropriate isotype controls.

All experiments were run with ECFC in passage 3 to 5 at 80–90% confluence. The concentrations of ethanol used in this study (0.5% (17 mM) and 1% (34 mM)) are doses that do not cause intoxication in vivo and correspond to 2 to 4 standard drinks.

2.2. In Vitro Angiogenesis Assay.We used anin vitroangio- genesis assay (endothelial tubule formation in Matrigel) to test the capacity of ECFC to form capillary tubule-like net- works. In a 96-well plate, 17,000 cells per well were seeded in 150μl treatment medium with 30μl growth factor reduced in Matrigel (BD Biosciences, Bedford, MA).

ECFCs were either treated with 0.5% and 1% ethanol for the duration of the assay or pretreated for 24 h with 0.5% and 1% ethanol. A corresponding control w/o etha- nol was run in tandem. After 16 h of incubation at 37°C and 5% CO2 supply, each well was photographed with a LEICA DMI 6000 B microscope. Total tubule length in each visual field was measured using the ImageJ software 1.52q (National Institutes of Health). All experiments were performed in triplicate wells from which values were averaged (n= number of experiments).

2.3. Cell Migration Assay.To analyze ECFC migratory ability, 50,000 ECFCs were seeded in each well of a 12-well plate with a growth medium containing 2.44% supplements, 5% FCS, and 1.2% penicillin/streptomycin. After reaching confluence, the ECFC monolayer was scratched using a sterile P200 pipette tip to produce a lane free of cells as described before [20]. ECFCs treated with 0.5% and 1% ethanol or after 24 h of preincubation with 0.5% or 1% ethanol and a correspond- ing control w/o ethanol were run in tandem. Light micro- scopic images were obtained immediately after the scratch (T0) and at the end of the experiment after 18 h (T18).

Migration into the scratch wound was analyzed using the ImageJ software and calculated as the percentage of wound closure (percentage of original area at T0 that became occu- pied by cells by migration into the wound area at T18). All

(3)

experiments were done in quadruplicate wells from which values were averaged.

2.4. Cell Impedance Assay.Cell impedance was determined by real-time impedance analysis using the xCelligence Real- time analyzer (RTCA, Roche, Mannheim, Germany). The Cell Index (CI), which reflects cell adherence, is converted from impedance measurement by the xCelligence software (Version 1.2.1) and was continuously monitored every 20 min for at least 72 h and directly after specific treatments of the cells. For experiments done, 10,000 cells were seeded in triplicates onto gold-coated E-Plate VIEW 96-well plates (Roche, Mannheim, Germany) and proliferation was calcu- lated through measuring increasing impedance. ECFCs treated with 0.5% and 1% ethanol or after 24 h of preincuba- tion with 0.5% or 1% ethanol and a corresponding control w/o ethanol were run in tandem.

2.5. Cell Proliferation Assay. To determine the proliferative capacity of ECFCs after treatment with ethanol, 50,000 cells were seeded per well of 6-well culture plates in EGM supple- mented with 5% (v/v) FBS and 1% penicillin/streptomycin.

After 24 h, 48 h, and 72 h of treatment, the cell number was counted in a Neubauer chamber with 1 : 2 trypan-blue dilu- tion. Population doubling time was calculated as the following:

log 2/ðlog ðNt/NoÞ/, t= time periodðhÞ, Nt = number of cells at timet, andNo = initial cell number. ECFCs after treat- ment with 0.5% EtOH 24 h and 1.0% EtOH and a corre- sponding untreated control were run in tandem.

2.6. Flow Cytometry.Flow cytometry analysis was performed to detect adhesion molecule expression on the ECFC surface.

ECFCs treated with 1% ethanol and a corresponding control w/o ethanol were run in tandem. Cells were harvested by incubating with Accutase (Capricorn, Ebsdorfergrund, Germany) for 10-15 min at room temperature. After washing with flow cytometry buffer (PBS, 2% BSA (Merck, Darmstadt, Germany)),1 × 105cells were blocked with nor- mal IgG (Grifols, Paris, France) (5 mg/ml) for 1 min, followed by incubation with the appropriate antibodies (VCAM1 APC (BioLegend, San Diego, CA), PECAM1 FITC (BioLegend, San Diego, CA), and VE-cadherin PE (BioLegend, San Diego, CA)) and isotype controls at 4°C for 30 minutes. For each experiment, a positive control for apoptotic cells was included to exclude dead cells from analysis. Apoptosis was induced by UV-irradiation with a transilluminator (Biostep, Jahnsdorf, Germany) for 30 min, and cells were further incubated for 2 h at room temperature. Cells were harvested, washed, and resuspended in Annexin V binding buffer, stained with Annexin V FITC (BioLegend, San Diego, CA) and incubated for 20 min. After washing and prior to measurement, propidium iodide (10μg/ml) (Sigma-Aldrich, Darmstadt, Germany) was added. Flow cytometry measure- ments were performed on a BD FACSCalibur Flow cyt- ometer (Becton Dickinson, Heidelberg, Germany), and results were analyzed using the FlowJo X Software V10 (Tree Star, Ashland, OR).

2.7. RT-PCR for Quantification of VEGF, sFLT-1, PlGF, and sEng Gene Expression.The RNA isolation of ECFC was per-

formed according to the protocol of Chomczynski et al.

which was slightly modified [21]. ECFCs treated with 1%

ethanol and a corresponding control w/o ethanol were run in tandem. The concentration of RNA of each sample was determined spectrophotometrically (BIO photometer, Eppendorf, Germany). For the cDNA synthesis, 2μg of RNA was diluted with diethylpyrocarbonate (DEPC) water to a volume of 8μl and denatured at 68°C for 10 min in a thermocycler (PTC 200, Biozym Scientific GmbH, Germany). Then, 12μl of High Capacity cDNA Reverse transcription (RT) master mix was added. In each case, 1.5μl diluted cDNA and 10.5μl master mix were pipetted into the appropriate strip tubes (0.1 ml). For each treat- ment, triplets were created and three RT-PCR runs were done for each patient. For normalization, 18S rRNA served as a housekeeping gene. Primer sequences used were as fol- lows: VEGF-A forward primer 5-TACCTCCACCATGCCA AGTG-3, VEGF-A reverse primer 5-GATGATTCTGCCCT CCTCCTT-3, sFlt1 forward primer 5-AATCAGAGGTG AGCACTGCAAC-3, sFlt1 reverse primer 5-TGGTAC AATCATTCCTTGTGCTTT-3, PlGF forward primer 5- CCTACGTGGAGCTGACGTTCT-3, PlGF reverse primer 5-CCTTTCCGGCTTCATCTTCTC-3, sEng forward primer 5-ACCTTTGGTGCCTTCCTCAT-3, sEng reverse primer 5-CAATCCCTCAGAGGCTTCAC-3, 18S rRNA forward primer 5-ACATCCAAGGAAGGCAGCAG-3, and 18S rRNA reverse primer 5-TTTTCGTCACTACCTCCCCG-3. Relative expression levels of ECFC from control vehicle and incuba- tion with ethanol werefinally compared.

2.8. Immunoblot for Quantification of VEGF, sFlt-1, and VE-Cadherin. For analysis of proteins VEGF, sFlt-1, and VE-cadherin, ECFCs were grown to 50% confluence in 100 mm dishes (Sarstedt, Nümbrecht, Germany) in an endo- thelial growth medium with 2.5% FCS with the respective additives. ECFCs were seeded into 100 mm dishes and treated with either only EGM (C = control) or EGM plus 1% ethanol. Cells were treated with 6 ml medium and cultivated for 24 h at 37°C, followed by harvesting of the cells, cell lysis, and protein quantification.

Protein lysates were separated by SDS-PAGE and transferred to nitrocellulose membranes (GE Healthcare, Braunschweig, Germany). After blocking for 1 h with 5%

dry milk powder in PBS-T, the membrane was incubated overnight at 4°C with PBS-T plus 5% dry milk powder and appropriate antibodies: 1 : 1000 anti-VEGF (Abcam, Cambridge, UK), 1 : 400 anti-sFlt-1 (Sigma-Aldrich, St. Louis, Missouri, USA), and 1 : 2000 anti-VE-cadherin (Abcam, Cambridge, UK). After three times of washing with PBS-T, the secondary antibody was added (1 : 5000 goat anti-rabbit;

GE Healthcare, Braunschweig, Germany) in 5% dry milk powder in PBS-T for 2 h at room temperature. Visualization of immunoblot bands was performed by using ECL chemilu- minescence (Pierce) and X-ray. The X-rays were scanned and analyzed with the ImageJ software.

2.9. ELISA for Quantification of Secreted Protein Levels of PlGF and sEng. PlGF and sEng concentrations were mea- sured by enzyme-linked immunosorbent assay (ELISA)

(4)

(PlGF & sEng Quantikine ELISA Kit R&D Systems, United States) according to the manufacturer’s recommendations.

Briefly, 100μl of concentrated cell culture supernatant for PlGF or 50μl for sEng was added per well. Standards and controls were assayed in duplicates and samples in triplicates and after incubation with the appropriate conjugates measured by using a microplate reader (Thermo Fisher Scientific) at 450 nm.

2.10. Statistical Analysis.The collected individual measured values (n) from experiments were analyzed with GraphPad Prism7 (GraphPad Software Inc.) for their statistical rele- vance with a Mann-WhitneyU-test or unpairedt-test after

determination of data distribution using the Shapiro Wilk normality test. Data are shown as mean and standard deviation (SD). A significant deviation of the value pairs is indicated by aPvalue (<0.05) and by one asterisk (∗).

3. Results

3.1. In Vitro Angiogenesis. A Matrigel angiogenesis model was used to assess the capacity of ECFC to differentiate into tubule-like structures (Figure 1). Tubule assemblage by ECFC pretreated for 24 h with ethanol was markedly higher in comparison to untreated ECFC (control:1:64∗107± 4:0

∗106μm; 0.5% ethanol: 2:17∗107± 4:4∗106μm, P=

Control

Control 0 𝜇m750

0 𝜇m750

0 𝜇m750

0 𝜇m750

0 𝜇m750

+0.5 % EtOH

+0.5 % EtOH

+0.5 % EtOH and preincubation

+1.0 % EtOH and preincubation

+1.0 % EtOH

+1.0 % EtOH +0.5 % EtOH

24h preincubation

+1.0 % EtOH

Total length of tubule formation (in 𝜇m)

6 7 7

Figure1: Eect of ethanol on capillary-tube formation in a Matrigel assay. ECFCs were cultured in endothelial basal medium ((EBM) +5%

FBS) and treated with 0.5% or 1% ethanol directly prior assay start or 24 h before or without ethanol (control). Capillary-tube formation (average total tubule length per microscopiceld) was analyzed after 16 h by visual microscopy at 2.5x magnication. Data are expressed as total tubule length inμm. Results represent mean values of total tubulelength ± SDof at least 6 independent experiments.P< 0:05vs.

untreated control.

(5)

0:001; 1.0% ethanol:2:01∗107± 4:5∗106μm,P= 0:03). In the absence of preincubation, ethanol did not impact tubule formation (0.5% ethanol: 1:66∗107± 5:2∗106μm, P= 0:36; 1.0% ethanol:1:84∗107± 4:3∗106μm,P= 0:48).

3.2. Migration.With scratch wound areafilling expressed as percent of total possible lane closure (100%), the migration of ECFC in the presence and absence of ethanol was tested (Figure 2). Migration by ethanol-treated ECFC was markedly higher in comparison to untreated control (control: 22:6%

± 8:7%; 0.5% ethanol: 31:4%± 11:5%, P= 0:03; 1.0%

ethanol:37:4%± 11:5%, P= 0:001; 24 h preincubation 0.5%

ethanol: 34:6%± 13:8%, P= 0:01; 24 h preincubation 1.0%

ethanol:29:8%± 8:8%,P= 0:004).

3.3. Cell Impedance.To compare the cell impedance of ECFC treated with ethanol and controls, changes in impedance were determined. Cell impedance of ethanol-treated ECFC was measured up to 72 h and was markedly higher in com- parison to untreated control (Pvalues at timepoint 72 h vs.

control: 0.5% ethanol:P= 0:004, 1% ethanol:P= 0:001; pre- incubation 0.5% ethanol:P= 0:01; preincubation 1% ethanol:

P= 0:64) (Figure 3).

3.4. Proliferation.The population doubling time of ECFC was shorter in ethanol-treated cells even though not significantly different (control: 24.91 h; 0.5% ethanol: 20.75 h;P= 0:43vs.

control, 1% ethanol: 17.42 h;P= 0:44vs. control) (Figure 4).

3.5. VE-Cadherin, VCAM, and PCAM Expression.Usingflow cytometry, we tested the ethanol-dependent cell surface expression of VE-cadherin, VCAM, and PCAM on ECFC (Figure 5). The expression of VE-cadherin by ethanol- treated ECFC was significantly higher in comparison to untreated control (control:3:77%± 3:1%; 1% ethanol: 27:9

%± 15:0%,P= 0:01) whereas VCAM and PCAM expression was not different (VCAM: control:3:2%± 2:2%; 1% ethanol:

14:0%± 10:1%, P= 0:31; PCAM: control: 97:3%± 1:0%; +1% ethanol:97:2%± 1:8%,P= 0:96).

3.6. VEGF, sFLT-1, PlGF, and sEng Gene Expression.There was no significant difference in the VEGF, sFLT-1, PlGF, or sEng gene expression of ECFC treated with ethanol in com- parison to vehicle control (Figure 6(a): VEGF: P= 0:60;

sFlt-1:P= 0:26; PlGF:P= 0:93; sEng:P= 0:96).

3.7. Protein Expression Level of VEGF, sFlt-1, VE-Cadherin, and Secreted Protein Levels of sEng and PlGF.There was no significant difference in the protein expression level of VEGF, sFlT-1, or VE-cadherin of ECFC treated with ethanol in comparison to vehicle control (Figure 6(b): VEGF:P= 0:99;

sFlT-1:P= 0:86; VE-cadherin:P= 0:75). Also, the secreted protein level of sEng and PlGF did not differ between ethanol-treated cells and vehicle control (Figure 6(c): sEng:

P= 0:13; PlGF:P= 0:13).

4. Discussion

In this study, we demonstrate a promoting effect of moderate ethanol concentrations on angiogenic capacities of ECFC. In

this context, we tested the ability of migration, proliferation, and tubule formation, which were enhanced after incubation with moderate concentrations of ethanol. These functional

15

Control +0.5 % EtOH

+0.5 % EtOH

+1.0 % EtOH

+1.0 % EtOH

+0.5 % EtOH

+0.5 % EtOH and 24h preincubation

+1.0 % EtOH and 24h preincubation

24h preincubation

T0

T0 0mm1

0mm1

0mm1

0mm1 0mm1

0mm1 0mm1

0mm1 0mm1

0mm1 T0

T0 T0 Control

T8

T8

T8

T8

T8

+1.0 % EtOH

% of migration

65 55 45 35 25

Figure 2: Eect of ethanol on ECFC migration. ECFCs were cultured in endothelial basal medium (EBM) +5% FBS and in the absence or presence of 0.5% or 1% ethanol directly prior assay start or 24 h before. The migration of ECFC into the scratch wound was assessed after incubation for 8 h. Results of at least 6 independent experiments represent mean ± SD percent wound lling.P< 0:05vs. untreated control.

(6)

properties represent cell characteristics important for angio- genesis and vasculogenesis, and they are markers of vascular health. Ethanol-treated cells showed a higher expression level of the cell surface marker VE-cadherin, a protein which plays a significant role in cell-cell interaction. Interestingly, the positive effect of ethanol seems not to be triggered by VEGF, PlGF, sEng, or sFlt-1. Additionally, expression levels of their mRNAs, protein, or soluble protein expression were not dif- ferent after ethanol treatment compared to untreated control.

Ourfindings confirm data of previous studies in which a stimulating effect of ethanol on endothelial function is dem- onstrated [26, 27]. To our knowledge, however, this is the first study to demonstrate enhanced functional properties

of ECFC treated with ethanol. ECFCs are an endothelial cell type with a strong intrinsic clonal proliferation potential and the ability to contribute to de novo blood vessel forma- tion in vitroandin vivo [21, 22]. The ECFC recruited into the damaged tissue was derived either from the circulation or from the local vascular wall [23]. The endothelial integrity of the vascular wall is restored by migration and proliferation of ECFC [24, 25]. Taken together, based on the current accumulated evidence, ECFCs are the most rational and promising cell sources that are able to incorporate into or form vessels directly in areas of tissue regeneration.

There are some epidemiological studies that demonstrate the benefits of moderate ethanol consumption in ischemic heart disease [5–7]. Interestingly, the results of these works suggest that in addition to lowering the rate of atherosclerosis through regular exposure to ethanol over many years, at least part of the ethanol-induced protection of the vascular endo- thelium can be induced within a few minutes by a short expo- sure. This protection can be provided by moderate ethanol doses, which correspond to about one or two alcoholic bever- ages. At this juncture, the concentrations of ethanol used in this study, i.e., 0.5% (17 mM) and 1% (34 mM), are levels that do not cause intoxication in vivo and correspond to 2 to 3 standard drinks per day [28]. Chen et al. showed that the treatment of cardiac muscle cells with 10-50 mM ethanol protects against ischemia by activating protein kinase C [26]. The ability of ethanol to activate protein kinase C has also been observed in several other cell systems [29, 30] and has been reported to mediate the protection of the heart from ischemia. In vivostudies demonstrated cardiovascular pro- tection after prolonged ethanol administration of 25-50 mM also in a guinea pig model.

Restoring blood flow after tissue injury or occlusion requires angiogenic germination of endothelial cells from

7.5 0

5 10 15

12.5 17.5 22.5

24 h 48 h 72 h

27.5 32.5 37.5 Time (in h)

Cell index

42.5 47.5 52.5 57.5 62.5 67.5

Control EtOH 0.5 % EtOH 1.0 %

EtOH preincubation 0.5 % EtOH preincubation 1.0 %

Figure3: Eect of ethanol on ECFC cell impedance. ECFCs were incubated in the absence or presence of ethanol (0.5% or 1% or 24 h of preincubation with 0.5% or 1% ethanol) in EGM +5% (v/v) FBS. Cell impedance was measured after 72 h for at least 6 independent experiments.

2.5

2.0

1.5

1.0

Number of counted viable cells

5.0

0

0 24 48 72

5

Figure4: Effect of ethanol on ECFC proliferation rate. ECFCs were incubated in the absence or presence of ethanol (0.5% or 1%

ethanol) in EGM +5% (v/v) FBS. Cell numbers were counted, and population doubling time was calculated after 24 h for at least 4 independent experiments.

(7)

nearby intact blood vessels, as well as vasculogenesis through circulating ECFC to allow invasion of new blood vessels to restore tissue perfusion. A short-term treatment with ethanol to increase the capacity of ECFC could be a possible thera- peutic approach to enable the bloodflow to be restored more quickly.

The exact reasons why ethanol can protect the cardiac tis- sue and endothelium have not yet been fully clarified. It is certain that ethanol can change the biophysical and biochem- ical properties of cell membranes. Ethanol interacts directly with membrane proteins to modulate their activity. In differ- ent cell models, exposure of ethanol impacted both the signal transduction mediated by protein kinase C [31, 32] and the cAMP-dependent protein kinase [30, 33].

In our study, we show an increased expression of the membrane surface molecule VE-cadherin, which is signifi- cant for cell-cell interaction. Processes, which are mediated by adhesion molecules, are activation of cell mobilization, migration, and proliferation via loosening of endothelial cell adhesion complexes. These mechanisms are impaired in CVD [17, 34]. Widner et al. showed that the expression of

VE-cadherin is reduced in plaque microvessels leading to vascular damage [35]. The observed increase of VE- cadherin in our study might contribute to an increased repair capacity of the endothelium.

With regard to the cardioprotective effect, half a million adults and 15000 children undergo open-heart surgery annu- ally in the United States each year in which the heart is exposed to controlled periods of ischemia. Despite advances in heart protection, myocardial dysfunction remains a major cause of morbidity and mortality in the immediate postoper- ative period. The exact time of the expected ischemia is known in this context. Therefore, the targeted use of a cardi- oprotective agent can prevent the consequences of ischemia in the endothelium and the remaining heart tissue. One could speculate that moderate doses of ethanol could be car- dioprotective and this approach could be evaluated further.

ECFCs seem to be a promising cell source for revasculo- genesis in damaged tissue in CVDs. They have excellent vas- cularization potentialin vitroandin vivo, and in this study, we show that their potential is further increased by adding small amounts of ethanol. ECFC can be isolated in sufficient

100

60 80

40

Frequence of total (in %)

20 0

PCAM

EtOH EtOH EtOH

VCAM VE - cadherin

(a)

PCAM

Counts

VE cadherin

CountsCounts

100 101 102 103 104

100 101 102 103 104

100 101 102 103 104

Negative control Isotype EtOH

Control

Negative control Isotype

EtOH Control

Negative control Isotype EtOH Control

(b)

Figure5: Ethanol-dependent ECFC cell surface expression of VE-cadherin, VCAM, and PECAM1. (a) ECFCs were treated with or without 1% ethanol and incubated with antibodies (anti-VE-cadherin, anti-VCAM, and anti-PECAM1) and analyzed byow cytometry. Expression levels of VCAM, VE-cadherin, and PCAM are shown. Results of at least 8 independent experiments represent cell surface expression in % shown asmean ± SD. P< 0:05vs. untreated control. (b) Flow cytometry analysis of PCAM, VCAM, and VE-cadherin on ECFC cell surface after treatment with EtOH.

(8)

numbers from umbilical cord blood for possible use in new- borns but if stored long-term could be used as an autologous source of reparative cells for the treatment of CVDs in adult- hood. In older children and adults, primarily circulating peripheral blood ECFCs are less clonogenic, proliferative, and angiogenic than umbilical cord ECFC. Finally, it is known that certain disease states such as diabetes can reduce the frequency and function of the isolated ECFC to such an extent that these cells are not of sufficient quality for use as revascularization therapy [36, 37]. Here, short-term therapy with ethanol could possibly increase the capacity and proliferation power of the ECFC in order to achieve a higher quantity and quality of these cells.

In conclusion, ethanol at moderate concentrations enhances the angiogenic abilities of ECFC. Ethanol-exposed cells showed a higher expression level of VE-cadherin, sug- gesting an increased endothelial repair capacity. Ourfindings support data from epidemiological studies demonstrating the benefits of short exposure of ethanol to prevent cardiovascu- lar damage in adults [5–7]. However, the data provided here are limited due to the use of primary cells underin vitrocon- ditions and show the short-term effect of ethanol on endo- thelial functionality. Further studies on the impact of long-

term moderate ethanol exposure on ECFC biology are still needed.

Data Availability

The experimental data and row data used to support the findings of this study are available from the corresponding author upon request.

Conflicts of Interest

No conflicts of interest,financial, or otherwise are declared by the authors.

Acknowledgments

We greatly appreciate the support of the faculty, residents, and staffof the Gynecology Research Unit and the Division of Obstetrics at Hannover Medical School in recruiting par- ticipants and collecting blood samples and Katja Borns for technical assistance. The study was supported by the German Research Foundation (DFG) Project Grant VE490/7-1 (to F.

von Versen-Höynck).

VEGF

EtOH EtOH EtOH EtOH

sFLT-1 PIGF s-ENG 3

–5

2

mRNA expression

1

0

(a)

2.0 1.5

1.0

Protein expression normalized to beta-actin 0.5

0.0

VEGF

EtOH EtOH EtOH

sFLT-1 VE - cadherin

(b) 800

600

400

Secreted protein level

200

0

s-ENG

EtOH EtOH

PIGF

(c)

150 kDa

Control EtOH

VE cadherin

sFLT-1 VEGF Beta-actin 100 kDa

50 kDa 37 kDa

(d)

Figure 6: Validation of ethanol-dependent expression of VEFG, PlGF, sFlt-1, sEng, and VE-cadherin. Gene expression levels, protein expression, and secreted protein levels of ECFC from untreated ECFC or after treatment with 1% ethanol were determined. For each experimental group, triplicates were created and RT-PCR runs (a), immunoblot (b, d), and ELISA (c) were performed for each ECFC cell line. Results representmean values ± SDof at least 6 independent experiments.

(9)

References

[1] T. Kurihara, H. Tomiyama, H. Hashimoto, Y. Yamamoto, E. Yano, and A. Yamashina, Excessive alcohol intake increases the risk of arterial stiening in men with normal blood pressure,Hypertension research : ocial journal of the Japanese Society of Hypertension, vol. 27, no. 9, pp. 669673, 2004.

[2] G. Fenelon, C. E. B. Balbão, R. Fernandes et al.,Characteriza- tion of the acute cardiac electrophysiologic effects of ethanol in dogs,Alcoholism, Clinical and Experimental Research, vol. 31, no. 9, pp. 15741580, 2007.

[3] L. Djousse, I. M. Lee, J. E. Buring, and J. M. Gaziano,Alcohol consumption and risk of cardiovascular disease and death in women: potential mediating mechanisms,” Circulation, vol. 120, no. 3, pp. 237244, 2009.

[4] A. L. Klatsky, “Alcohol and cardiovascular diseases,” Expert Review of Cardiovascular Therapy, vol. 7, no. 5, pp. 499506, 2014.

[5] J. R. Parratt,Protection of the heart by ischaemic precondi- tioning: mechanisms and possibilities for pharmacological exploitation,” Trends in Pharmacological Sciences, vol. 15, no. 1, pp. 1925, 1994.

[6] L. O. de Labry, R. J. Glynn, M. R. Levenson, J. A. Hermos, J. S.

LoCastro, and P. S. Vokonas,Alcohol consumption and mor- tality in an American male population: recovering the U- shaped curve–findings from the normative Aging Study, Journal of Studies on Alcohol, vol. 53, no. 1, pp. 25–32, 1992.

[7] S. K. Ahlawat and S. B. Siwach,“Alcohol and coronary artery disease, International Journal of Cardiology, vol. 44, no. 2, pp. 157162, 1994.

[8] H. Kobayashi, M. Ashraf, P. M. Rahamathulla, and M. Minami,Moderating eect of low doses of ethanol on reoxygenation injury in the anoxic myocardium,Pathology, Research and Practice, vol. 182, no. 6, pp. 810–816, 1987.

[9] T. W. Sheehy,“Alcohol and the heart,”Postgraduate Medicine, vol. 91, no. 5, pp. 271277, 2016.

[10] J. B. Hoek, R. Rubin, and A. P. Thomas,“Ethanol-induced phospholipase C activation is inhibited by phorbol esters in isolated hepatocytes, The Biochemical Journal, vol. 251, no. 3, pp. 865871, 1988.

[11] J. E. Dildy-Mayeld and R. A. Harris,Ethanol inhibits kainate responses of glutamate receptors expressed in Xenopus oocytes: role of calcium and protein kinase C, The Journal of neuroscience : the ocial journal of the Society for Neurosci- ence, vol. 15, no. 4, pp. 31623171, 1995.

[12] T. F. King and J. H. McDermott,Endothelial progenitor cells and cardiovascular disease,Journal of Stem Cells, vol. 9, no. 2, pp. 93–106, 2014.

[13] N. Werner, S. Kosiol, T. Schiegl et al.,“Circulating endothelial progenitor cells and cardiovascular outcomes, The New England Journal of Medicine, vol. 353, no. 10, pp. 9991007, 2005.

[14] P. I. Sipos, I. P. Crocker, C. A. Hubel, and P. N. Baker,Endo- thelial progenitor cells: their potential in the placental vascula- ture and related complications,”Placenta, vol. 31, no. 1, pp. 1–

10, 2010.

[15] F. L. Celletti, J. M. Waugh, P. G. Amabile, A. Brendolan, P. R.

Hilker, and M. D. Dake,Vascular endothelial growth factor enhances atherosclerotic plaque progression, Nature Medi- cine, vol. 7, no. 4, pp. 425–429, 2001.

[16] S. Sankaralingam, I. A. Arenas, M. M. Lalu, and S. T. Davidge,

“Preeclampsia: current understanding of the molecular basis of vascular dysfunction,Expert Reviews in Molecular Medicine, vol. 8, no. 3, pp. 120, 2006.

[17] Y. Yan, Q. Chang, Q. Li et al.,Identication of plasma vascu- lar endothelia-cadherin as a biomarker for coronary artery dis- ease in type 2 diabetes mellitus patients,”International Journal of Clinical and Experimental Medicine, vol. 8, no. 10, pp. 1946619470, 2015.

[18] P. E. Ronksley, S. E. Brien, B. J. Turner, K. J. Mukamal, and W. A. Ghali, Association of alcohol consumption with selected cardiovascular disease outcomes: a systematic review and meta-analysis,BMJ, vol. 342, no. feb22 1, p. d671, 2011.

[19] M. Grundmann, M. Haidar, S. Placzko et al., Vitamin D improves the angiogenic properties of endothelial progenitor cells, American journal of physiology Cell physiology, vol. 303, no. 9, pp. C954C962, 2012.

[20] S. A. Bainbridge, J. M. Roberts, F. von Versen-Hoynck, J. Koch, L. Edmunds, and C. A. Hubel,Uric acid attenuates tropho- blast invasion and integration into endothelial cell mono- layers, American journal of physiology Cell physiology, vol. 297, no. 2, pp. C440C450, 2009.

[21] D. Tasev, P. Koolwijk, and V. W. M. van Hinsbergh,Thera- peutic potential of human-derived endothelial colony- forming cells in animal models, Tissue engineering Part B, Reviews, vol. 22, no. 5, pp. 371–382, 2016.

[22] Y. Lin, D. J. Weisdorf, A. Solovey, and R. P. Hebbel,“Origins of circulating endothelial cells and endothelial outgrowth from blood,The Journal of Clinical Investigation, vol. 105, no. 1, pp. 7177, 2000.

[23] T. Asahara, A. Kawamoto, and H. Masuda,Concise review:

circulating endothelial progenitor cells for vascular medicine, Stem Cells, vol. 29, no. 11, pp. 1650–1655, 2011.

[24] D. A. Ingram, L. E. Mead, D. B. Moore, W. Woodard, A. Fenoglio, and M. C. Yoder,Vessel wall-derived endothelial cells rapidly proliferate because they contain a complete hierarchy of endothelial progenitor cells, Blood, vol. 105, no. 7, pp. 2783–2786, 2005.

[25] L. Hubert, R. Darbousset, L. Panicot-Dubois et al., Neutro- phils recruit and activate human endothelial colony-forming cells at the site of vessel injury via P-selectin glycoprotein ligand-1 and L-selectin,Journal of thrombosis and haemosta- sis : JTH, vol. 12, no. 7, pp. 11701181, 2014.

[26] C. H. Chen, M. O. Gray, and D. Mochly-Rosen,Cardiopro- tection from ischemia by a brief exposure to physiological levels of ethanol: role of epsilon protein kinase C,Proceedings of the National Academy of Sciences of the United States of America, vol. 96, no. 22, pp. 1278412789, 1999.

[27] L. Vergori, E. Lauret, R. Soleti, M. C. Martinez, and R. Andriantsitohaina, Low concentration of ethanol favors progenitor cell differentiation and neovascularization in high-fat diet-fed mice model, The International Journal of Biochemistry & Cell Biology, vol. 78, pp. 4351, 2016.

[28] S. Moore, J. Shepherd, N. Perham, and B. Cusens,The preva- lence of alcohol intoxication in the night-time economy, Alcohol and Alcoholism, vol. 42, no. 6, pp. 629634, 2007.

[29] B. Hundle, T. McMahon, J. Dadgar, C. H. Chen, D. Mochly- Rosen, and R. O. Messing,“An inhibitory fragment derived from protein kinase Cepsilon prevents enhancement of nerve growth factor responses by ethanol and phorbol esters,The Journal of Biological Chemistry, vol. 272, no. 23, pp. 15028 15035, 1997.

(10)

[30] R. O. Messing,Ethanol as an enhancer of neural dierentia- tion,” Alcohol and Alcoholism. Supplement, vol. 2, pp. 289–

293, 1993.

[31] I. Diamond, B. Wrubel, W. Estrin, and A. Gordon,Basal and adenosine receptor-stimulated levels of cAMP are reduced in lymphocytes from alcoholic patients, Proceedings of the National Academy of Sciences of the United States of America, vol. 84, no. 5, pp. 14131416, 1987.

[32] P. L. Homan, P. Valverius, M. Kwast, and B. Tabako,Com- parison of the eects of ethanol on beta-adrenergic receptors in heart and brain, Alcohol and Alcoholism. Supplement, vol. 1, pp. 749–754, 1987.

[33] D. M. Lovinger and Q. Zhou,Alcohols potentiate ion current mediated by recombinant 5-HT3RA receptors expressed in a mammalian cell line,” Neuropharmacology, vol. 33, no. 12, pp. 15671572, 1994.

[34] G. Favero, C. Paganelli, B. Buoli, L. F. Rodella, and R. Rezzani,“Endothelium and Its Alterations in Cardiovascu- lar Diseases: Life Style Intervention, BioMed Research International, vol. 2014, Article ID 801896, 28 pages, 2014.

[35] R. J. Widmer and A. Lerman,“Endothelial dysfunction and cardiovascular disease,Global cardiology science & practice, vol. 2014, no. 3, pp. 291308, 2014.

[36] E. K. Blue, R. DiGiuseppe, E. Derr-Yellin et al.,“Gestational diabetes induces alterations in the function of neonatal endo- thelial colony-forming cells, Pediatric Research, vol. 75, no. 2, pp. 266272, 2014.

[37] Y. P. R. Jarajapu, S. Hazra, M. Segal et al.,Vasoreparative dysfunction of CD34+ cells in diabetic individuals involves hypoxic desensitization and impaired autocrine/paracrine mechanisms,”PLoS ONE, vol. 9, no. 4, p. e93965, 2014.

Referenzen

ÄHNLICHE DOKUMENTE

In summary, whereas methylene blue stimulates the oxidation of ethanol to CO 2 in isolated hepatocytes and intact rats, no effect of methylene blue on the disposition of ethanol and

As low as 0.1 µg/mL (Caki1 and KTCTL-26) or 0.2 µ g/mL curcumin (A498) significantly reduced tumor cell growth following irradiation, while 0.1 or 0.2 µ g/mL curcumin alone or

The plots show the quantitative analysis as a function of the position in the ET axis with respect to area and circumference for the individual entities cartilage, lumen, OFP, TVPM

The coordination sphere of the silver atoms is further complemented by an ethanol molecule which is also engaged in hydrogen bonding with one of the sulfonate oxygen atoms..

The effects of the nitric oxide synthase inhibitor 7-nitroindazole on ethanol pharmacokinetics in rats after acute and chronic ethanol administration.. Nitric oxide synthase

In this study, we further discovered a positive rela- tionship between oocyte maturation rate and FF NT-4, which, together with the dynamics expression of TRKB and NT-4 in mice

There- fore, the copyright of the article has been changed to © The Author(s) 2020 and the article is forthwith distributed under the terms of the Creative Commons Attribution

The “Ethanol Expansion Coalition” assembling the Ministério da Agri- cultura, Pecuária e Abastecimento (Ministry of Agriculture, Livestock and Food Supply), Ministério do